9
Histone deacetylase inhibitor SAHA induces ERa degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling Xin Yi, Wei Wei, Sheng-Yu Wang, Zhi-Yan Du, Yuan-Ji Xu, Xiao-Dan Yu * Department of Pathology, Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China 1. Introduction Breast cancer is the most common form of malignant disease in woman worldwide. It has been reported that ERa plays a critical role in the initiation and progression of breast cancer because approximately 70% of primary breast cancers are ERa positive [1,2]. As a result, ERa has become an important target in the treatment of hormone-responsive breast cancer. Unfortunately, most patients initially responding to anti- estrogen therapies, such as tamoxifen, which is a standard component of front-line therapy for ERa positive breast cancer, will eventually become resistant to those therapies [3]. Aromatase inhibitors (AIs) are new drugs used for endocrine treatment of post-menopausal breast cancer and have demonstrated efficacy in patients with breast cancer resistant to anti-estrogens. However, resistance to AIs has also been observed. The potential mechanisms of endocrine resistance are not fully understood, but evidence suggests biochemical pharmacology 75 (2008) 1697–1705 article info Article history: Received 12 August 2007 Accepted 31 October 2007 Keywords: HDACi SAHA ERa Breast cancer CHIP Hsp90 abstract Estrogen receptor a (ERa) plays an important role in the development and progression of breast cancer, and recent studies showed that ERa expression is associated with resistance to hormonal therapy. Therefore, a number of studies have explored ways to deplete ERa from breast cancer cells as a new therapy especially for hormone-refractory breast cancer. We reported here that suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, effectively depletes ERa in breast cancer MCF-7 cells. However, the intrinsic mechanisms by which SAHA decreases ERa levels are not clear. Our present data demon- strated that both inhibition of ERa mRNA level and promotion of ERa degradation by the proteasome contribute to SAHA-induced ERa depletion, indicating that SAHA may exert its effects through transcriptional and posttranslational mechanisms. Furthermore, the decrease of ERa protein level in MCF-7 cells after SAHA treatment is mainly the result of its rapid degradation by the ubiquitin-proteasome pathway rather than transcriptional inhibition. In addition, we showed that inactivation of the heat shock protein-90 (Hsp90) is involved in SAHA-induced ERa degradation, and ubiquitin ligase CHIP (C-terminal Hsc70 interacting protein) enhances SAHA-induced ERa degradation. SAHA-induced ERa depletion is paralleled with reduction of transcriptional activity of ERa and SAHA is able to effectively inhibit cell proliferation and induce apoptosis of MCF-7 cells. Taken together, our results revealed a mechanism for SAHA-induced ERa degradation and indicated that SAHA is a suitable pharmacological agent for depletion of ERa and a potential choice for breast cancer expressing high ERa. # 2007 Elsevier Inc. All rights reserved. * Corresponding author. Tel.: +86 10 6693 2372; fax: +86 10 6821 3039. E-mail address: [email protected] (X.-D. Yu). available at www.sciencedirect.com journal homepage: www.elsevier.com/locate/biochempharm 0006-2952/$ – see front matter # 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.bcp.2007.10.035

Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

  • Upload
    xin-yi

  • View
    218

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 5

avai lab le at www.sc iencedi rec t .com

journal homepage: www.e lsev ier .com/ locate /b iochempharm

Histone deacetylase inhibitor SAHA induces ERa degradationin breast cancer MCF-7 cells by CHIP-mediated ubiquitinpathway and inhibits survival signaling

Xin Yi, Wei Wei, Sheng-Yu Wang, Zhi-Yan Du, Yuan-Ji Xu, Xiao-Dan Yu *

Department of Pathology, Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China

a r t i c l e i n f o

Article history:

Received 12 August 2007

Accepted 31 October 2007

Keywords:

HDACi

SAHA

ERa

Breast cancer

CHIP

Hsp90

a b s t r a c t

Estrogen receptor a (ERa) plays an important role in the development and progression of

breast cancer, and recent studies showed that ERa expression is associated with resistance

to hormonal therapy. Therefore, a number of studies have explored ways to deplete ERa

from breast cancer cells as a new therapy especially for hormone-refractory breast cancer.

We reported here that suberoylanilide hydroxamic acid (SAHA), a histone deacetylase

inhibitor, effectively depletes ERa in breast cancer MCF-7 cells. However, the intrinsic

mechanisms by which SAHA decreases ERa levels are not clear. Our present data demon-

strated that both inhibition of ERa mRNA level and promotion of ERa degradation by the

proteasome contribute to SAHA-induced ERa depletion, indicating that SAHA may exert its

effects through transcriptional and posttranslational mechanisms. Furthermore, the

decrease of ERa protein level in MCF-7 cells after SAHA treatment is mainly the result of

its rapid degradation by the ubiquitin-proteasome pathway rather than transcriptional

inhibition. In addition, we showed that inactivation of the heat shock protein-90 (Hsp90) is

involved in SAHA-induced ERa degradation, and ubiquitin ligase CHIP (C-terminal Hsc70

interacting protein) enhances SAHA-induced ERa degradation. SAHA-induced ERa depletion

is paralleled with reduction of transcriptional activity of ERa and SAHA is able to effectively

inhibit cell proliferation and induce apoptosis of MCF-7 cells. Taken together, our results

revealed a mechanism for SAHA-induced ERa degradation and indicated that SAHA is a

ical agent for depletion of ERa and a potential choice for breast cancer

expressing high ERa.

suitable pharmacolog

# 2007 Elsevier Inc. All rights reserved.

1. Introduction

Breast cancer is the most common form of malignant disease

in woman worldwide. It has been reported that ERa plays a

critical role in the initiation and progression of breast cancer

because approximately 70% of primary breast cancers are ERa

positive [1,2]. As a result, ERa has become an important target

in the treatment of hormone-responsive breast cancer.

Unfortunately, most patients initially responding to anti-

* Corresponding author. Tel.: +86 10 6693 2372; fax: +86 10 6821 3039.E-mail address: [email protected] (X.-D. Yu).

0006-2952/$ – see front matter # 2007 Elsevier Inc. All rights reserveddoi:10.1016/j.bcp.2007.10.035

estrogen therapies, such as tamoxifen, which is a standard

component of front-line therapy for ERa positive breast

cancer, will eventually become resistant to those therapies

[3]. Aromatase inhibitors (AIs) are new drugs used for

endocrine treatment of post-menopausal breast cancer and

have demonstrated efficacy in patients with breast cancer

resistant to anti-estrogens. However, resistance to AIs has also

been observed. The potential mechanisms of endocrine

resistance are not fully understood, but evidence suggests

.

Page 2: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 51698

that tamoxifen-resistant tumors regrowth may be associated

with ERa signaling. Moreover, complex interactions between

ERa and growth factor signaling pathways are also involved,

which is thought to be one of the determinants of endocrine

resistance [4–6]. Therefore, depletion of ERa from breast

cancer cells may be a particularly powerful approach to block

ERa signaling, especially ERa/growth factor crosstalk, pre-

venting the development of endocrine resistance ultimately.

Unliganded ERa, like other steroid hormone receptors, is

maintained in a ligand-binding competent conformation by

associating with various Hsp90-based chaperone complexes

[7]. Hsp90 inhibitors, such as geldanamycin (GA), can inhibit

Hsp90 molecular chaperone function and induce ERa degra-

dation through the ubiquitin-proteasome pathway [8,9].

Recently, it was found that carboxyl terminus of Hsc70-

interacting protein (CHIP), which is a co-chaperone of Hsp90/

Hsp70 complex, is involved in GA-induced ERa degradation as

an ubiquitin ligase [10]. However, anti-estrogen fulvestrant,

which also induces ERa degradation by dissociating the

complex of Hsp90 with ERa, reducing the interaction between

CHIP and ERa after treatment with GA, indicating that distinct

downstream pathways exist for ERa degradation by treatment

with different drugs [10]. Therefore, it is necessary for us to

elucidate the molecular mechanism of new drug-induced-ERa

degradation.

Histone deacetylase (HDAC) inhibitors, a promising class

of antitumor agents, can block the proliferation and induce

cell death in a wide variety of transformed cells [11].

However, the antitumor mechanism of HDAC inhibitors

has not been completely elucidated. It has been proved that

HDAC inhibitors selectively affect gene transcription by

acetylation of histones, such as SAHA and LAQ824, which

transcriptionally up-regulate p21 and increase p27 expres-

sion. This is associated with cell cycle arrest and apoptosis in

cancer cells, and is shown to induce in vivo regression of

tumors [12–14]. HDAC inhibitors-induced mitotic defect by

causing aberrant acetylation of histones in heterochromatin

and centromere domains per se can result in cell death by

either apoptosis or mitotic death/catastrophe [15]. It has also

been shown that SAHA can induce polyploidy and lead to cell

senescence in transformed cells [16]. These findings indi-

cated that HDAC inhibitors can cause tumor cell growth

arrest, apoptosis, mitotic cell death and polyploidy to exert

antitumor effects.

Recently, a number of studies showed that the antitumor

effects of HDAC inhibitors may be associated with Hsp90

acetylation induced by HDAC inhibitors, such as depsipeptide

(FK228), LAQ824 and SAHA. It has been proposed that Hsp90

acetylation correlates with inactivation of Hsp90 and induces

Hsp90 client proteins disassociation from Hsp90 molecular

complex, e.g. Raf-1, androgen receptor (AR) and HER2 [17–19].

An earlier research suggested that HDAC inhibitors deplete

ERa protein [20]. However, little is known regarding the

molecular mechanism of ERa depletion by HDAC inhibitors.

Suberoylanilide hydroxamic acid (SAHA) is an HDAC inhibitor,

which is in phase I/II clinical trials and has shown antitumor

activity in hematologic and solid tumors at doses well

tolerated by patients [21,22]. In present studies, we showed

that SAHA can suppress ERa mRNA level and induce ERa

degradation in breast cancer MCF-7 cells. The molecular

mechanism of SAHA-induced ERa degradation is associated

with inactivation of Hsp90, and co-chaperone CHIP of Hsp90 is

also involved as E3 ubiquitin ligase. The effect of SAHA on ERa

degradation is augmented by CHIP overexpression, but not

CHIP mutants’ overexpression, suggesting that CHIP partici-

pates in SAHA-induced ERa degradation. Most importantly,

SAHA-induced ERa depletion results in down-regulation of

ERa transcriptional activity and inhibition of cells proliferation

in MCF-7 cells. Therefore, SAHA may exert its antitumor effect

via depletion of ERa and inactivation of the crosstalk between

ERa and growth receptors signaling pathways.

2. Materials and methods

2.1. Cell, reagents and plasmids

The breast cancer cell line, MCF-7 cell line was obtained from

American Type Culture Collection (Manassas, VA, USA).

Suberoylanilide hydroxamic acid (SAHA) was offered by

AstraZeneca Company (Macclesfield, SK, UK). 17b-Estradiol,

4-hydroxytamoxifen, and MG-132 were purchased from Sigma

Chemical Company (St. Louis, MO, USA). ICI 182,780 was

purchased from Tocris Cookson Ltd. (Ellisville, MO, USA).

Antibodies for Raf-1, phospho-AKT, AKT, phospho-ERK, ERK,

acetylated-lysine, survivin, Myc, actin and ubiquitin were

obtained from Cell Signaling Technology (Beverly, MA, USA).

Anti-ERa and anti-CDK4 antibody was purchased from Santa

Cruz Biotechnology (Santa Cruz, CA, USA). Anti-Hsp90 anti-

body and anti-Hsp70 antibody were obtained from Stressgen

Biotechnologies Corporation (Victoria, BC, Canada). Anti-CHIP

antibody was purchased from Abcam Ltd. (Cambridge, UK).

Lipofectamine 2000 was obtained from Invitrogen Corporation

(Rockville, MD, USA). The Myc-CHIP (WT), Myc-CHIP (DTPR),

and Myc-CHIP (DU-box) constructs were kindly provided by Dr.

Yili Yang (NIH/NCI). ERE-pS2-Luc was kindly offered by Dr. Qi-

Nong Ye (Beijing Institute of Biotechnology, Beijing, China).

2.2. Cell culture and transient transfection

MCF-7 cells were cultured in DMEM medium containing 10%

fetal bovine serum at 37 8C in 5% CO2. Before experiments,

cells were cultured in hormone-free medium (phenol red-free

MEM with 5% charcoal-stripped FBS) for 3 days. For transient

transfection, cells were transfected with an equal amount of

total plasmid DNA by using Lipofectamine 2000 according to

the manufacturer’s guidelines.

2.3. Stable transfection

MCF-7 cells were transfected with pcDNA3.1-Myc-CHIP (WT),

pcDNA3.1-Myc-CHIP (DTPR), pcDNA3.1-Myc-CHIP (DU-box), or

empty vector by using Lipofectamine 2000 and selected in

growth medium containing 800 mg/ml G418 for 3 weeks. Then

drug-resistant colonies were chosen and expanded in growth

medium containing 300 mg/ml G418. The expression of Myc-

CHIP (WT), Myc-CHIP (DTPR), and Myc-CHIP (DU-box) in stable

cell lines (MCF-7) were detected by Western blot with anti-Myc

antibody. In parallel, several empty plasmid-transfected

clones were randomly selected and used as control cells.

Page 3: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 5 1699

2.4. Western blot, immunoprecipitation analysis andluciferase assay

For Western blot analysis, control or SAHA-treated MCF-7

cells were lysed in Laemmli buffer (Bio-Rad Laboratories, CA,

USA), approximately 60 mg of total proteins were resolved on

SDS polyacrylamide gels and immunoblot was performed as

previously described [19]. For immunoprecipitation experi-

ments, cellular extracts from approximately 1 � 107 cells

were prepared in RIPA buffer, approximately 400 mg of total

proteins were used for immunoprecipitation analysis fol-

lowed by the procedure previously described [19]. For

luciferase assay, cell lysates were prepared and carried out

as recommended by Luciferase Assay System (Promega). Data

were expressed as mean � S.E.M. Statistical analysis was

performed by Student’s t-test and P values less than 0.05 were

considered significant.

2.5. RT-PCR

Total RNA was extracted from cultured cells with TRIzol

reagent according to the manufactured guidelines (Invitro-

gen). Reverse transcription (RT) was performed using First

Strand cDNA Synthesis Kit (Fermentas). Primer pairs used for

ERa PCR were as follows: forward, 50-TGATCCTACCA-

GACCCTTCA-30, and reverse, 50-TCCTGTCCAAGAGCAAGTT-

30. G3PDH was amplified and used as a standard for the PCR

reaction.

Fig. 1 – SAHA induces ERa degradation via the ubiquitin-protea

with different concentrations of SAHA for 24 h or treated with 5

the protein level of ERa was examined with anti-ERa antibody b

indicated times in the presence of proteasome inhibitor MG-13

extracted and mRNA level of ERa was detected by RT-PCR analy

treated with an inhibitor of protein synthesis, 100 mM cyclohexim

at indicated times. As a control, MCF-7 cells were treated with

protein level of ERa was detected by Western blot with anti-ERa

4 h in the presence of proteasome inhibitor MG-132 or in the ab

30 min before and continued during the SAHA treatment. Cell ly

with anti-ERa antibody, resolved by SDS-PAGE, and immunoblo

treated with SAHA (5 mM) at indicated times in the presence of

(5 mM). As a control, MCF-7 cells were treated with MG-132 (5 m

detected by Western blot with anti-ERa antibody.

2.6. Cell proliferation assay

MCF-7 cells were plated into 96-well plates at a density of

6 � 104/well and cultured in hormone-free medium. After 3

days, the medium was replaced with fresh medium in the

presence or absence of 1.25–10 mM SAHA. At appropriate time

points, the percentages of viable cells after treatment were

measured using the MTT assay. Each experiment was

performed three times.

3. Results

3.1. SAHA depletes ERa via the ubiquitin-proteasomepathway in MCF-7 cells

To investigate the effect of SAHA on ERa expression, we used

SAHA to treat breast cancer MCF-7 cells, which express high

ERa protein, the results showed that SAHA induces dose- and

time-dependent inhibition of ERa protein level (Fig. 1A).

Similar inhibition was observed with depsipeptide (FK228)

and trichostatin A (data not shown), which are type I and pan-

histone deacetylase inhibitors, respectively [20]. Previous

reports showed that HDAC inhibitors like LAQ824 exert their

effects on depleting androgen receptor through transcrip-

tional and posttranslational mechanisms [18]. To determine

whether SAHA-mediated ERa depletion is due to the same

mechanisms, we treated MCF-7 cells with SAHA for 6–24 h and

some pathway in MCF-7 cells. (A) MCF-7 cells were treated

mM SAHA at indicated times. Cell lysate was prepared and

y Western blot. (B) MCF-7 cells were treated with SAHA at

2 or in the absence of MG-132 (5 mM). Total RNA was

sis, G3PDH served as loading control. (C) MCF-7 cells were

ide (CHX) for 30 min, followed by addition of SAHA (5 mM)

100 mM cycloheximide (CHX) for 2, 4 and 6 h alone. The

antibody. (D) MCF-7 cells were treated with SAHA (5 mM) for

sence of MG-132 (5 mM). MG-132 was added to the cells

sate was lysed in RIPA buffer and immunoprecipitated (IP)

tted (IB) with anti-ubiquitin antibody. (E) MCF-7 cells were

proteasome inhibitor MG-132 or in the absence of MG-132

M) for 2, 4 and 6 h alone. The level of ERa protein was

Page 4: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 51700

performed RT-PCR analysis to detect ERa mRNA level.

Comparison of the declining speed of ERa in protein and in

mRNA levels showed that, the remarkable decline of ERa

protein appeared at 6 h followed by the SAHA exposure,

whereas the faint decline of ERa in mRNA level appeared until

SAHA treatment for 12 h (Fig. 1B). The lagging of transcrip-

tional inhibition suggested that SAHA may decline ERa protein

level mainly via affecting its stability. To prove this hypoth-

esis, we treated MCF-7 cells for various times with SAHA, in

the presence or absence of cyclohexamide (CHX), an inhibitor

of protein synthesis, in the presence or absence of SAHA, and

then measured the relative ERa protein level in MCF-7 cells. As

shown in Fig. 1C, ERa protein level decreases faster in the cells

treated with SAHA in the presence of CHX than in the cells

treated with CHX alone. This result supported that inhibitory

effects of SAHA on ERa are mainly due to accelerate ERa

degradation in MCF-7 cells. To determine whether the

ubiquitin-proteasome system is responsible for SAHA-

induced ERa degradation, we treated MCF-7 cells with SAHA

at indicated times in the presence or absence of proteasome

inhibitor MG-132. ERa protein was immunoprecipitated and its

ubiquitination status was examined by Western blot with

anti-ubiquitin antibody. Fig. 1D shows that SAHA treatment

induces ubiquitination of ERa and this effect is evidently

enhanced by cotreatment of SAHA and MG-132. Furthermore,

Fig. 2 – SAHA acetylates Hsp90 and induces dissociation of ERa

proteins in MCF-7 cells. (A) SAHA induces Hsp90 acetylation. MC

lysed in RIPA buffer. Acetylated lysine was immunoprecipitated

PAGE, and immunoblotted (IB) with anti-Hsp90 or anti-ERa ant

loading control. (B) SAHA disassociates the binding between ERa

indicated times, cell lysate was lysed in RIPA buffer and immuno

PAGE, and immunoblotted (IB) with anti-Hsp90 or anti-ERa anti

as the loading control. (C) SAHA depletes Hsp90 client proteins.

the cell lysate from MCF-7 cells treated with SAHA (5 mM) for 6–

the proteasome inhibitor MG-132 prevents SAHA-induced ERa

degradation (Fig. 1E), suggesting that SAHA could induce ERa

degradation via the ubiquitin-proteasome pathway. Although

proteasome inhibitor MG-132 can completely prevent SAHA-

induced ERa protein degradation, it has no effect on SAHA-

induced decline on ERa mRNA level (Fig. 1B).

3.2. SAHA induces Hsp90 acetylation and Hsp90-ERacomplex disassociation in MCF-7 cells

It has been discovered that the association of ERa with the

heat shock protein-90 (Hsp90) molecular chaperone complex

is required for its stability and function [7]. Recently, it was

shown that HDAC inhibitors induce acetylation of Hsp90,

which inhibits its ATP binding and chaperone association with

its client proteins [17–19]. Consistent with these studies, we

next determined whether Hsp90 acetylation is involved in

SAHA-induced ERa depletion. Further experiments were then

performed to assess the effect of SAHA on acetylation of Hsp90

and ERa by immunoprecipitation with anti-acetylated lysine

antibody and immunoblotting with anti-Hsp90 or anti-ERa

antibodies, respectively. The results showed that treatment

with 5 mM SAHA as short as 2 h has already induced

acetylation of Hsp90 in MCF-7 cells, and the effect was

enhanced at 4 h but without significantly affecting the protein

from Hsp90 chaperone complex and depletes Hsp90 client

F-7 cells, treated with SAHA (5 mM) at indicated times, were

(IP) with anti-acetylated lysine antibody, resolved by SDS-

ibody. The level of Hsp90 in cell lysate was used as the

and Hsp90. MCF-7 cells were treated with SAHA (5 mM) at

precipitated (IP) using anti-ERa antibody, resolved by SDS-

body. The levels of Hsp90 and ERa in cell lysate were used

Western blot analyses of Raf-1, CDK4, survivin and actin in

48 h.

Page 5: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 5 1701

level of Hsp90 in MCF-7 cells. Interestingly, in the same

conditions acetylation of ERa is not observed (Fig. 2A). Next, we

estimated the effect of SAHA on the association of Hsp90-ERa.

Following treatment with SAHA for 2 or 4 h, Hsp90-ERa

coimmunoprecipitation assay was done. As shown in Fig. 2B,

SAHA induces the disassociation of Hsp90 and ERa paralleled

by increased acetylation of Hsp90. These data implied that

SAHA induces Hsp90 acetylation and Hsp90-ERa disassocia-

tion in MCF-7 cells, resulting in ERa degradation via the

ubiquitin-proteasome pathway. Previous studies showed that

inactivation of Hsp90 molecular chaperone could induce

depletion of its client proteins such as AKT, Raf-1, and steroid

receptors [23–25]. We further evaluated the levels of Hsp90

client proteins such as Raf-1, CDK4 and survivin. Fig. 2C shows

that SAHA also mediates the depletion of these Hsp90 clients.

These results further indicated that SAHA-induced Hsp90

acetylation leads to disruption of its chaperone function and

decrease of its client proteins.

3.3. CHIP participates in SAHA-induced ERa degradationas an ubiquitin ligase

Previous studies showed that carboxyl-terminus of HSC70-

interecting protein (CHIP) is a U-box-containing E3 ubiquitin

ligase that binds through its tetratricopeptide repeat (TPR)

domain to independent TPR acceptor sites on Hsp90 and

Hsp70 [26,27]. CHIP has been shown to facilitate the

ubiquitination of Hsp90 client proteins, such as p53 and ErbB2

[28,29]. Most recently, CHIP has been reported to play a role in

GA-induced degradation of ERa [10]. Whether CHIP also plays a

role in SAHA-induced ERa degradation has not been investi-

gated. To determine whether CHIP participates in SAHA-

induced ERa degradation, we transfected wild type CHIP and

deficient mutant plasmids (DTPR and DU-box) into MCF-7 cells

and established the stable transfected cell lines. As a control,

MCF-7-Mock (MCF-7 cells stably transfected with empty

vector) was also established. We treated MCF-7-Mock cells

and MCF-7-CHIP (WT) with SAHA at indicated times. Fig. 3A

shows that ERa degradation is evidently accelerated in MCF-7-

CHIP (WT) cells compared with MCF-7-Mock cells. It has been

reported that CHIP associates with ERa and promotes ERa

ubiquitination in MCF-7 cells, we wonder to investigate the

function of CHIP in the case of SAHA-induced ERa degradation.

We treated MCF-7-CHIP (WT) cells with SAHA for 2 or 4 h, ERa

protein was immunoprecipitated with anti-ERa antibody, and

coimmunoprecipitated CHIP was detected by immunoblotting

with anti-Myc antibody. As shown in Fig. 3B, SAHA enhances

association of CHIP with ERa in a time-dependent manner. We

next examined whether CHIP could act as an ubiquitin ligase

mediating SAHA-induced ERa degradation. MCF-7-Mock cells

and MCF-7-CHIP (WT) cells were treated with SAHA for 2 h,

respectively, ubiquitination of ERa was slightly increased in

MCF-7-CHIP (WT) cells treated with SAHA (Fig. 3C), suggesting

a role for CHIP in SAHA-induced ERa ubiquitination. There-

fore, these results suggested that CHIP, by facilitating ERa

ubiquitination, targets ERa for proteasome-mediated degra-

dation. Furthermore, we evaluated whether overexpression of

the deletion mutant CHIP including CHIP (DTPR) and CHIP (DU-

box) could exert E3 ubiquitin ligase function in SAHA-induced

ERa degradation. As shown in Fig. 3D, overexpression of CHIP

(DU-box) and CHIP (DTPR) mutant, but not CHIP (WT), have no

effects on SAHA-induced ERa degradation. These data

indicated that intact CHIP is necessary to mediate SAHA-

induced ERa degradation.

3.4. SAHA down-regulates ERa transcriptional activityand blocks survival signaling in MCF-7 cells

We have confirmed that SAHA is capable to depleting ERa

expression through two mechanisms involving inhibition of

ERa mRNA level and induction of ERa degradation via CHIP-

mediated ubiquitin-proteasome pathway. To determine

whether SAHA-induced ERa depletion is associated with

attenuated ERa transcriptional activity, following treatment

with SAHA for 6 h, we transiently transfected MCF-7 cells with

an ERE-pS2-Luc reporter plasmid, then stimulated with E2 for

24 h and analyzed with luciferase assay. As shown in Fig. 4A,

SAHA decreases E2-induced ERa transcriptional activity in a

dose dependent manner accompanied with remarkably

decline of ERa protein level at same time. We assume that

the inhibition effect of SAHA on ERa transcriptional activity is

indirectly caused by ERa protein degradation, more experi-

ments using stable transfected cell lines, MCF-7-CHIP (WT),

MCF-7-CHIP (DTPR) and MCF-7-CHIP (DU-box) further demon-

strated that SAHA causes a stronger decrease of ERa

transcriptional activity inMCF-7-CHIP (WT) cells but not in

MCF-7-CHIP (DTPR) and MCF-7-CHIP (DU-box) cells, which is

consistent with its effect on depleting ERa from different cell

lines (Fig. 3D).

We further evaluated the effects of SAHA on survival

signaling pathways in MCF-7 cells. As shown in Fig. 4B,

treatment with SAHA resulted in the reduction of phosphory-

lated ERK or phosphorylated AKT in a time-dependent

manner, indicating that ERK and AKT activities are inhibited

in these cells. Coincidence with the inhibition of ERK and AKT

activities, the level of cleaved-PARP was increased, suggesting

that inhibition of the Raf-1/MEK/ERK and PI3K/AKT survival

signaling pathways is involved in the SAHA-induced apopto-

sis. Next, MCF-7 stable transfected cells were treated with

SAHA for 24 h; the result showed that CHIP overexpression

enhances the effect of SAHA on inactivating survival signaling

pathways (Fig. 4B).

We have showed that treatment with 5 mM SAHA is able to

induce ERa degradation and inhibit ERa transcriptional

activity. To examine the growth inhibitory effect of SAHA in

MCF-7 cells, MCF-7 cells were treated with SAHA for 0–72 h.

MTT assay demonstrated that SAHA causes dose and time-

dependent decrease in cell viability. The viability of MCF-7

cells was inhibited by 60% after 72 h, suggesting that SAHA is

effective in inhibiting MCF-7 cell proliferation (Fig. 4C).

Furthermore, to evaluate the correlation between SAHA-

induced cytotoxicity and ERa depletion in MCF-7 cells, we

compared the growth inhibitory effect of SAHA on MCF-7 cells

with that on MCF-7-CHIP (WT), MCF-7-CHIP (DTPR) and MCF-7

(DU-box) cells. The result showed that the cytotoxicity of SAHA

is obviously enhanced in MCF-7-CHIP (WT) cells, whereas no

difference among MCF-7 cells, MCF-7-CHIP (DTPR) and MCF-7-

CHIP (DU-box) cells. It is indicated that promoting ERa

depletion by CHIP overexpression could enhance SAHA

cytotoxicity (Fig. 4C). Taken together, SAHA down-regulates

Page 6: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

Fig. 3 – CHIP is required for SAHA-induced ERa degradation in MCF-7 cells. (A) Overexpression of CHIP promotes SAHA-

induced ERa degradation. MCF-7 cells or MCF-7-CHIP (WT) cells were treated with SAHA (5 mM) at indicated times, cell

lysate was prepared and the protein levels of ERa and CHIP were detected by Western blot with anti-ERa and anti-Myc

antibody, respectively. (B) SAHA enhances CHIP-ERa interaction. MCF-7-CHIP (WT) cells were treated with SAHA (5 mM) at

indicated times, and cell lysate was lysed in RIPA buffer and immunoprecipitated (IP) using anti-ERa antibody, resolved by

SDS-PAGE, and immunoblotted (IB) with anti-ERa or anti-Myc antibody. (C) CHIP enhances SAHA-induced ERa

ubiquitination. Immunoprecipitation (IP) was performed with anti-ERa antibody and immunoblotted (IB) with anti-

ubiquitin, anti-ERa and anti-Myc antibody, respectively in the RIPA buffer from MCF-7 or MCF-7-CHIP (WT) cells treated

with SAHA (5 mM) for 2 h. The level of ERa in cell lysate was used as the loading control. (D) CHIP’s function is dependent on

both TPR and U-box domain. SAHA (5 mM) treated these stable transfected cells (MCF-7-Mock, MCF-7-CHIP (WT), MCF-7-

CHIP (DTPR) and MCF-7-CHIP (DU-box)) for 6 h. The expression of exogenesis CHIP and its mutants were detected by anti-

Myc antibody. ERa protein was detected by Western blot with anti-ERa.

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 51702

ERa transcriptional activity and blocks survival signaling in

MCF-7 cells via CHIP-mediated mechanism.

4. Discussion

It has been proposed that levels of ERa are commonly

increased in premalignant and malignant breast lesions,

and ERa plays a crucial role for the growth and survival of

breast cancer cells. Recently, accumulated evidences also

have shown that complex interactions between ERa and

growth factor signaling pathways are one of the potential

mechanisms of endocrine resistance to breast cancer [4–6].

Thus, ERa becomes an important target for the treatment of

breast cancer. Depletion of ERa protein by Hsp90 inhibitors or

ERa-small interfering RNA (siRNA) has been shown to

suppress breast tumor growth in vivo [8,9,30]. Therefore,

depletion of ERa from breast cancer cells is a rational

therapeutic strategy not only for primary estrogen-dependent

breast cancer but also for hormone-refractory breast cancer.

Our experiments confirmed that SAHA, a HDAC inhibitor,

currently in human clinical trials, is able to deplete ERa in

breast cancer MCF-7 cells.

Our data showed that SAHA depletes ERa through two

mechanisms: attenuation of its mRNA level and promotion

of its degradation by ubiquitin-proteasome pathway. It is

similar to what has been previously observed that HDAC

inhibitor LAQ824 and TSA deplete HER2, Bcr-Abl, and AR

through transcriptional and posttranslational mechanisms.

It seems that HDAC inhibitors-induced degradation of

oncoproteins maybe commonly accompanied by down-

regulation of their mRNA levels. However, recent report

showed that TSA has no effect on the levels of HIF-1a mRNA

transcripts while it induces rapid degradation of HIF-1a

protein [31], suggesting that the effect of HDAC inhibitors is

different for various types of Hsp90 client proteins. Our

current study has focused on the molecular mechanism of

SAHA-induced ERa degradation. Previous report showed

that TSA, another HDAC inhibitor, induced proteasome-

independent down-regulation of ERa [32]. However, in this

study, we observed that MG-132, a proteasome inhibitor,

could effectively inhibit SAHA-induced ERa degradation and

enhance ERa ubiquitination. Therefore, our data indicated

that SAHA induces ERa degradation via the ubiquitin-

proteasome pathway. The basis for these disparate results

is not clear, but may reflect different effects of TSA versus

Page 7: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

Fig. 4 – SAHA down-regulates ERa transcriptional activity and inactivates survival signaling in MCF-7 cells. (A) After

treatment with SAHA (5–10 mM) for 6 h, ERE-Luc plasmid (250 ng) was transiently transfected into MCF-7 cells and the stable

transfected cells (MCF-7-CHIP (WT), MCF-7-CHIP (DTPR) and MCF-7-CHIP (DU-box)) for 24 h; cells were treated with 10 nM E2

or DMSO as indicated and assayed 24 h later for relative luciferase activity. P < 0.01 (student’s t-test, vs. first group) is

indicated by a *. ERa protein was detected by Western blot with anti-ERa. (B) Left figure: MCF-7 cells were treated with SAHA

(5 mM) at indicated times. Right figure: MCF-7 cells and the stable transfected cells (MCF-7-CHIP (WT), MCF-7-CHIP (DTPR)

and MCF-7-CHIP (DU-box)) were treated with SAHA (5 mM) for 48 h. Cell lysate was analyzed by Western blot with

antibodies to p-AKT, AKT, p-ERK, ERK, PARP, Myc or actin. (C) Upper figure: MCF-7 cells were treated with SAHA at 1.25–

10 mM at indicated times. Lower figure: MCF-7 cells and the stable transfected cells (MCF-7-CHIP (WT), MCF-7-CHIP (DTPR)

and MCF-7-CHIP (DU-box)) were treated with SAHA at 1.25–10 mM for 48 h. Cytotoxicity was evaluated by MTT assays. All

experiments were performed in triplicate.

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 5 1703

SAHA or attribute to the different doses and times of MG-132

used in the studies.

Recently it has been showed that HDAC inhibitors such as

LAQ824, FK228 and SAHA exert their anti-cancer effects by

inactivating of Hsp90 molecular chaperone function, impair

the chaperone association of Hsp90 with its client proteins,

HER2, AR and induce the degradation via the ubiquitin-

proteasome pathway. In our studies, the results indicated that

SAHA-induced ERa degradation in MCF-7 cells is also

associated with inactivation of Hsp90 molecular chaperone

function. First, treatment with SAHA induces Hsp90 acetyla-

tion, and Hsp90 acetylation is linked to the inactivation of its

chaperone function. The disassociation of ERa from the Hsp90

chaperone complex is accompanied with SAHA-induced

acetylation of Hsp90. Secondly, besides ERa, other Hsp90

client proteins, such as Raf-1, CDK4 and survivin are also

reduced by SAHA. Therefore, these observations suggested

that SAHA induces ERa degradation via the ubiquitin-protea-

some pathway by inhibiting Hsp90 chaperone function in

breast cancer MCF-7 cells. Previous reports have shown that

treatment with SAHA induces p21 and cell cycle growth arrest,

associated with differentiation and apoptosis of breast cancer

cells [13]. Our data indicated that antitumor effects of SAHA

are also associated with inhibition of Hsp90 function, which

leads to Hsp90 client proteins’ degradation.

It is known that molecular chaperones recognize non-

native proteins and aid in their correct folding. When it is

unsuccessful, the misfolded proteins will be directed to

degradation. The mutually exclusive pathways of folding

and degradation constitute the cell’s protein quality control

Page 8: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 51704

system [33]. As recently studies showed, another co-chaper-

one, CHIP, regulates chaperone function in part by regulating

ubiquitin-proteasome pathway and determining whether

proteins enter the productive folding pathway or the degrada-

tion pathway. CHIP acts as an E3 ubiquitin ligase and promotes

ubiquitination of Hsp90 client proteins by a COOH-terminal U-

box domain, while binding molecular chaperone Hsp90 or

Hsp70 through TPR domain [34]. Recently it has been reported

that ERa serves as a substrate for Hsp90/Hsp70-associated

CHIP. The association of CHIP with ERa through a chaperone

intermediately results in ubiquitination and degradation of

ERa. And the presence of Hsp90 inhibitor GA potently

stimulates this process. Our data demonstrated that E3

ubiquitin ligase CHIP also mediates SAHA-induced ERa

ubiquitination and proteasomal degradation. CHIP overex-

pression in the presence of SAHA leads to an additive loss of

ERa and decreases its transcriptional activity via enhanced

ERa ubiquitination. The accelerate degradation of ERa further

mediates inactivation of survival signaling pathways and

enhances SAHA-induced cytotoxicity. The function of CHIP on

mediating ERa degradation is dependent on both the TPR

domain and the U-box E3 ligase activity, as transfection with

either CHIP (DTPR) or CHIP (DU-box) has no effect on SAHA-

induced ERa degradation or cytotoxicity. Taken together, we

concluded that E3 ubiquitin ligase CHIP is necessary for ERa

degradation, following Hsp90 inhibition by SAHA. These

findings are consistent with previous studies, which showed

that overexpression of CHIP by transient transfection

enhances ERa degradation [10].

Although tamoxifen has been the standard endocrine

treatment for breast cancer for many years, its extended use

may be associated with the development of tamoxifen

resistance [3]. The potential mechanisms of endocrine

resistance are not fully clarified, but evidence suggested that

complex interactions between ERa and growth factor signaling

pathways are involved. The introduction of new endocrine

agents that can induce ERa depletion and block growth

signaling pathways, may improve the therapeutic options for

women with endocrine-resistant breast cancer. Here, we

demonstrated that SAHA, a HDAC inhibitor, induces rapid

depletion of ERa protein, resulting in repression of the ERa-

dependent transcriptional function. Consistent with our

results, Reid and his colleagues reported that another HDAC

inhibitor, TSA, has the similar effect on influencing estrogen-

dependent transcription [35]. However, their data showed that

TSA-induced ERa clearance is due to reduction of the steady-

state level of ERa mRNA in MCF-7 cells and consequently

suppress the transactivation activity of ERa. In contrast, our

results showed at 6 h of SAHA treatment, the protein level of

ERa decreased markedly accompanied with inhibition of ERa-

dependent transcriptional activation, but with slight down-

regulation of ERa mRNA level, suggesting that SAHA-induced

ERa transcriptional inactivation is mainly caused by ERa

degradation, but not reduction of ERa mRNA. Our results

implied that inhibition of ERa-dependent transcriptional

activation by SAHA could provide a potentially important

strategy to develop new therapeutic agents against breast

cancer. Activation of EGFR and HER2 receptors results in

stimulation of several signaling pathways including mitogen-

activated protein kinase (MAPK) and PI3K/AKT pathways. It

has been reported that ERa can be phosphorylated and

activated by these protein kinases; increased bidirectional

crosstalk between ERa and EGFR signaling pathways is

thought to contribute to the development of tamoxifen

resistance [6]. In our study, we demonstrated that SAHA

could reduce MAPK and PI3K/AKT signaling activity and

inhibit the growth of MCF-7 cells. Our data indicated that the

use of agents such as SAHA to degrade ERa protein level and

abrogate survival signaling pathways may lead to improve-

ments in the prevention of endocrine resistance. Our findings

also indicated that CHIP can mediate down-regulation of ERa

transcriptional activity and inhibition of survival signaling in

MCF-7 cells caused by SAHA, which implied that CHIP may

play an important role in HDAC inhibitors-induced antitumor

effects.

Acknowledgements

We thank Dr. Yili Yang and Dr. Qi-Nong Ye for providing

plasmids. This work was supported by the National Natural

Science Foundation of China (Grant Nos. 30330620 and

30470898).

r e f e r e n c e s

[1] Allred DC, Mohsin SK. Biological features of premalignantdisease in the human breast. J Mammary Gland BiolNeoplasia 2000;5(4):351–64.

[2] Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogenreceptor status by immunohistochemistry is superior to theligand-binding assay for predicting response to adjuvantendocrine therapy in breast cancer. J Clin Oncol1999;17(5):1474–81.

[3] Ali S, Coombes RC. Endocrine-responsive breast cancer andstrategies for combating resistance. Nat Rev Cancer2002;2(2):101–12.

[4] Lonning PE. Cross-resistance to different aromataseinhibitors in breast cancer treatment. Endocr Relat Cancer1999;6(2):251–7.

[5] Gutierrez MC, Detre S, Johnston S, Mohsin SK, Shou J, AllredDC, et al. Molecular changes in tamoxifen-resistant breastcancer: relationship between estrogen receptor, HER-2, andp38 mitogen-activated protein kinase. J Clin Oncol2005;23(11):2469–76.

[6] Levin ER. Bidirectional signaling between the estrogenreceptor and the epidermal growth factor receptor. MolEndocrinol 2003;17(3):309–17.

[7] Pratt WB, Toft DO. Steroid receptor interactions with heatshock protein and immunophilin chaperones. Endocr Rev1997;18(3):306–60.

[8] Beliakoff J, Bagatell R, Paine-Murrieta G, Taylor CW,Lykkesfeldt AE, Whitesell L. Hormone-refractory breastcancer remains sensitive to the antitumor activity of heatshock protein 90 inhibitors. Clin Cancer Res2003;9(13):4961–71.

[9] Bagatell R, Khan O, Paine-Murrieta G, Taylor CW, Akinaga S,Whitesell L. Destabilization of steroid receptors by heatshock protein 90-binding drugs: a ligand-independentapproach to hormonal therapy of breast cancer. ClinCancer Res 2001;7(7):2076–84.

[10] Fan M, Park A, Nephew KP. CHIP (carboxyl terminus ofHsc70-interacting protein) promotes basal and

Page 9: Histone deacetylase inhibitor SAHA induces ERα degradation in breast cancer MCF-7 cells by CHIP-mediated ubiquitin pathway and inhibits survival signaling

b i o c h e m i c a l p h a r m a c o l o g y 7 5 ( 2 0 0 8 ) 1 6 9 7 – 1 7 0 5 1705

geldanamycin-induced degradation of estrogen receptor-alpha. Mol Endocrinol 2005;19(12):2901–14.

[11] Marks PA, Richon VM, Miller T, Kelly WK. Histonedeacetylase inhibitors. Adv Cancer Res 2004;91:137–68.

[12] Johnstone RW, Licht JD. Histone deacetylase inhibitors incancer therapy: is transcription the primary target? CancerCell 2003;4(1):13–8.

[13] Richon VM, Sandhoff TW, Rifkind RA, Marks PA. Histonedeacetylase inhibitor selectively induces p21WAF1expression and gene-associated histone acetylation. ProcNatl Acad Sci USA 2000;97(18):10014–9.

[14] Fuino L, Bali P, Wittmann S, Donapaty S, Guo F, YamaguchiH, et al. Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells totrastuzumab, taxotere, gemcitabine, and epothilone B. MolCancer Ther 2003;2(10):971–84.

[15] Cimini D, Mattiuzzo M, Torosantucci L, Degrassi F. Histonehyperacetylation in mitosis prevents sister chromatidseparation and produces chromosome segregation defects.Mol Biol Cell 2003;14(9):3821–33.

[16] Xu WS, Perez G, Ngo L, Gui CY, Marks PA. Induction ofpolyploidy by histone deacetylase inhibitor: a pathway forantitumor effects. Cancer Res 2005;65(17):7832–9.

[17] Yu X, Guo ZS, Marcu MG, Neckers L, Nguyen DM, Chen GA,et al. Modulation of p53, ErbB1, ErbB2, and Raf-1 expressionin lung cancer cells by depsipeptide FR901228. J Natl CancerInst 2002;94(7):504–13.

[18] Chen L, Meng S, Wang H, Bali P, Bai W, Li B, et al. Chemicalablation of androgen receptor in prostate cancer cells bythe histone deacetylase inhibitor LAQ824. Mol Cancer Ther2005;4(9):1311–9.

[19] Bali P, Pranpat M, Swaby R, Fiskus W, Yamaguchi H, BalasisM, et al. Activity of suberoylanilide hydroxamic Acidagainst human breast cancer cells with amplification ofher-2. Clin Cancer Res 2005;11(17):6382–9.

[20] Margueron R, Duong V, Bonnet S, Escande A, Vignon F,Balaguer P, et al. Histone deacetylase inhibition andestrogen receptor alpha levels modulate the transcriptionalactivity of partial antiestrogens. J Mol Endocrinol2004;32(2):583–94.

[21] Kelly WK, Richon VM, O’Connor O, Curley T, MacGregor-Curtelli B, Tong W, et al. Phase I clinical trial of histonedeacetylase inhibitor: suberoylanilide hydroxamic acidadministered intravenously. Clin Cancer Res 2003;9(10 Pt1):3578–88.

[22] Kelly WK, O’Connor OA, Krug LM, Chiao JH, Heaney M,Curley T, et al. Phase I study of an oral histone deacetylaseinhibitor, suberoylanilide hydroxamic acid, in patients withadvanced cancer. J Clin Oncol 2005;23(17):3923–31.

[23] Fortugno P, Beltrami E, Plescia J, Fontana J, Pradhan D,Marchisio PC, et al. Regulation of survivin function byHsp90. Proc Natl Acad Sci USA 2003;100(24):13791–6.

[24] Schulte TW, Blagosklonny MV, Ingui C, Neckers L.Disruption of the Raf-1-Hsp90 molecular complex results indestabilization of Raf-1 and loss of Raf-1-Ras association. JBiol Chem 1995;270(41):24585–8.

[25] Stepanova L, Leng X, Parker SB, Harper JW. Mammalianp50Cdc37 is a protein kinase-targeting subunit of Hsp90 thatbinds and stabilizes Cdk4. Genes Dev 1996;10(12):1491–502.

[26] Connell P, Ballinger CA, Jiang J, Wu Y, Thompson LJ,Hohfeld J, et al. The co-chaperone CHIP regulates proteintriage decisions mediated by heat-shock proteins. Nat CellBiol 2001;3(1):93–6.

[27] Cyr DM, Hohfeld J, Patterson C. Protein quality control: U-box-containing E3 ubiquitin ligases join the fold. TrendsBiochem Sci 2002;27(7):368–75.

[28] Esser C, Scheffner M, Hohfeld J. The chaperone-associatedubiquitin ligase CHIP is able to target p53 for proteasomaldegradation. J Biol Chem 2005;280(29):27443–8.

[29] Zhou P, Fernandes N, Dodge IL, Reddi AL, Rao N, Safran H,et al. ErbB2 degradation mediated by the co-chaperoneprotein CHIP. J Biol Chem 2003;278(16):13829–37.

[30] Fu HJ, Jia LT, Bao W, Zhao J, Meng YL, Wang CJ, et al. Stableknockdown of estrogen receptor alpha by vector-basedRNA interference suppresses proliferation and enhancesapoptosis in breast cancer cells. Cancer Biol Ther2006;5(7):842–7.

[31] Kong X, Lin Z, Liang D, Fath D, Sang N, Caro J. Histonedeacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1 alpha. Mol Cell Biol 2006;26(6):2019–28.

[32] Alao JP, Lam EW, Ali S, Buluwela L, Bordogna W, Lockey P,et al. Histone deacetylase inhibitor trichostatin A repressesestrogen receptor alpha-dependent transcription andpromotes proteasomal degradation of cyclin D1 in humanbreast carcinoma cell lines. Clin Cancer Res2004;10(23):8094–104.

[33] Wickner S, Maurizi MR, Gottesman S. Posttranslationalquality control: folding, refolding, and degrading proteins.Science 1999;286(5446):1888–93.

[34] McDonough H, Patterson C. CHIP: a link between thechaperone and proteasome systems. Cell StressChaperones 2003;8(4):303–8.

[35] Reid G, Metivier R, Lin CY, Denger S, Ibberson D, Ivacevic T,et al. Multiple mechanisms induce transcriptional silencingof a subset of genes, including oestrogen receptor alpha, inresponse to deacetylase inhibition by valproic acid andtrichostatin A. Oncogene 2005;24(31):4894–907.